Therapy of Osteoarthritis: Comparison
Please note this is a comparison between Version 2 by Enzi Gong and Version 3 by Enzi Gong.

Osteoarthritis (OA) is a complex, multifactorial degenerative disease of the joint, characterized by chronic inflammation, progressive loss of articular cartilage, subchondral bone sclerosis and osteophyte formation, changes in the synovial membrane and increased volume of synovial fluid with altered coefficient of friction. In some respects, it can also be viewed as an inflammatory disease, leading to chronic pain and decrease of life quality.

  • liposomes
  • osteoarthritis
  • intra-articular
  • polysaccharides
  • polyphenols
  • anti-inflammatory activity

1. Overview

Osteoarthritis (OA) is a degenerative joint disease. An objective of the nanomedicine and drug delivery systems field is to design suitable pharmaceutical nanocarriers with controllable properties for drug delivery and site-specific targeting, in order to achieve greater efficacy and minimal toxicity, compared to the conventional drugs. The aim of this review is to present recent data on natural bioactive compounds with anti-inflammatory properties and efficacy in the treatment of OA, their formulation in lipid nanostructured carriers, mainly liposomes, as controlled release systems and the possibility to be intra-articularly (IA) administered. The literature regarding glycosaminoglycans, proteins, polyphenols and their ability to modify the cell response and mechanisms of action in different models of inflammation are reviewed. The advantages and limits of using lipid nanoformulations as drug delivery systems in OA treatment and the suitable route of administration are also discussed. Liposomes containing glycosaminoglycans presented good biocompatibility, lack of immune system activation, targeted delivery of bioactive compounds to the site of action, protection and efficiency of the encapsulated material, and prolonged duration of action, being highly recommended as controlled delivery systems in OA therapy through IA administration. Lipid nanoformulations of polyphenols were tested both in vivo and in vitro models that mimic OA conditions after IA or other routes of administration, recommending their clinical application. 

2. Osteoarthritis

Osteoarthritis (OA) is a complex, multifactorial degenerative disease of the joint, characterized by chronic inflammation, progressive loss of articular cartilage, subchondral bone sclerosis and osteophyte formation, changes in the synovial membrane and increased volume of synovial fluid with altered coefficient of friction [1][2][3][4][5][6]. In some respects, it can also be viewed as an inflammatory disease, leading to chronic pain and decrease of life quality [5][7][8]. During OA progression, the degradation process of the collagen network takes place constantly and a variety of inflammatory mediators are detected in the articular cartilage. Tumor necrosis factor alpha (TNF-α) and interleukin-1β (IL-1β) influence chondrocyte metabolism, and also induce the production of inflammatory mediators, such as nitric oxide (NO), and prostaglandin E2 [9]. In these conditions, cartilage is further degraded and the inflammatory process is perpetuated [1][10][11]. Several studies indicated that local joint inflammation (synovitis) induced by endogenous molecular products derived from cellular stress and extracellular matrix degradation acted through innate inflammatory network and could influence the integrity and function of articular cartilage [12][13]. On the other hand, the systemic inflammation resulting from metabolic disturbance could also contribute to OA progression [13][14]. Some reports presented OA as a systemic disease and described the complexity of the involved inflammatory mechanisms [15]. Currently, there are no efficient treatments that can stop the pathological processes involved in OA progression, but prevention strategies and treatments directed to symptoms, pain relieve and function regain [16][17]. Treatments are based on various pharmacologic agents, such as selective cyclooxygenase-2 (COX-2) inhibitors, non-steroidal anti-inflammatory drugs (NSAIDs), corticosteroids, even analgesics [18]. Their administration through oral route involves limited bioavailability and risk of side effects, such as upper gastrointestinal and cardiovascular complications [19]. As OA has a localized nature, intra-articular (IA) administration of drugs provides the opportunity to improve the treatment by local depot formation and prolonged drug action [13][20][21][22][23][24]. Although numerous disease-modifying OA drugs (DMOADs) showed promising results in preclinical trials, their poor IA bioavailability limited the treatment approval [25]. In the last years, natural bioactive molecules (e.g., glycosaminoglycans (GAGs) from animal sources or plant polyphenols) have gained considerable interest as therapeutic alternatives [19][26][27][28][29][30][31][32][33][34][35]. However, the efficacy of different anti-inflammatory bioactive molecules administration is limited due to their poor stability in the harmful biological milieu or low solubility, which decreases their bioavailability. Several delivery systems, including liposomes, microparticles, nanoparticles and hydrogels have been investigated for the sustained delivery and controlled release of bioactive molecules in the joints [5][36][37]. In vitro studies have demonstrated that in the case of OA, the phospholipidic layer acting as a boundary lubricant was missing from the articular surface of osteoarthritic degenerated cartilage and the structure of chondroitin sulfate (CS) was also changed [6]. Liposomes are the most commonly used nanocarriers to deliver drugs to human tissues in clinical applications and have been approved by the US Food and Drug Administration (FDA) [38]. As drug carrier systems, liposomes possess many biophysical and physicochemical properties suitable for IA administration, such as sustained release, ability for self-assembly and capacity to load large quantities of drugs [23][37]. Additionally, due to their ability to incorporate hydrophilic and hydrophobic molecules, good biocompatibility, low toxicity, activation and targeted delivery of bioactive compounds to the site of action, liposomes offer many advantages, such as the protection and efficiency of encapsulated material, solubilization of lipophilic molecules, prolongation of the duration of action and present targeting options. The clinical development of liposome-based drug delivery systems with synergistic therapeutic effect and a description of the technologies for NSAIDs liposomal formulations for orthopedic field applications were previously reviewed [39][40][41][42]. The only product approved in Germany and available on the market for IA administration in human patients with OA is Lipotalon®, containing the liposomal formulation of dexamethasone-21-palmitate [43].

3. Conclusions

Over recent years, delivery systems of biologically active compounds have become more advanced and complex when designing their lipid nanoformulation. Current research activities point towards finding an optimal formulation with suitable properties, capable of delivering encapsulated compounds. Liposomes have shown many advantages as carriers, including increased stability, reduced degradation, enhanced solubility of the drug, and improved pharmacokinetics. Several types of lipid nanocarriers have been developed for the treatment of different diseases, including OA, with increased attention being given to improve the delivery, efficacy, and safety. As we described in this review, lipid nanoformulations, mainly liposomes, loaded with natural bioactive molecules with anti-inflammatory activity, such as polysaccharides, polyphenols and proteins showed increased solubility and bioavailability, being able to improve their therapeutic effect in vivo. These are important aspects for further clinical research and the application of lipid nanoformulations for the treatment of OA.

References

  1. Sandell, L.J.; Aigner, T. Articular cartilage and changes in arthritis. An introduction: Cell biology of osteoarthritis. Arthritis Res. 2001, 3, 107–113.
  2. Bonnet, C.S.; Walsh, D.A. Osteoarthritis, angiogenesis and inflammation. Rheumatology 2005, 44, 7–16.
  3. Bottini, M.; Bhattacharya, K.; Fadeel, B.; Magrini, A.; Bottini, N.; Rosato, N. Nanodrugs to target articular cartilage: An emerging platform for osteoarthritis therapy. Nanomed. Nanotechnol. Biol. Med. 2016, 12, 255–268.
  4. Mora, J.C.; Przkora, R.; Cruz-Almeida, Y. Knee osteoarthritis: Pathophysiology and current treatment modalities. J. Pain Res. 2018, 11, 2189–2196.
  5. Kou, L.F.; Xiao, S.Y.; Sun, R.; Bao, S.H.; Yao, Q.; Chen, R.J. Biomaterial-engineered intra-articular drug delivery systems for osteoarthritis therapy. Drug Deliv. 2019, 26, 870–885.
  6. Jin, G.Z. Current Nanoparticle-Based Technologies for Osteoarthritis Therapy. Nanomaterials 2020, 10, 2368.
  7. Ma, X.; Zhang, Z.; Shen, M.; Ma, Y.; Li, R.; Jin, X.; Gao, L.; Wang, Z. Changes of type II collagenase biomarkers on IL-1β-induced rat articular chondrocytes. Exp. Ther. Med. 2021, 21, 582.
  8. Poulet, B.; Beier, F. Targeting oxidative stress to reduce osteoarthritis. Arthritis Res. Ther. 2016, 18.
  9. van den Bosch, M.H.J. Inflammation in osteoarthritis: Is it time to dampen the alarm(in) in this debilitating disease? Clin. Exp. Immunol. 2019, 195, 153–166.
  10. Mathiessen, A.; Conaghan, P.G. Synovitis in osteoarthritis: Current understanding with therapeutic implications. Arthritis Res. Ther. 2017, 19.
  11. Rahmati, M.; Mobasheri, A.; Mozafari, M. Inflammatory mediators in osteoarthritis: A critical review of the state-of-the-art, current prospects, and future challenges. Bone 2016, 85, 81–90.
  12. Liu-Bryan, R. Synovium and the Innate Inflammatory Network in Osteoarthritis Progression. Curr. Rheumatol. Rep. 2013, 15.
  13. Salgado, C.; Jordan, O.; Allémann, E. Osteoarthritis In Vitro Models: Applications and Implications in Development of Intra-Articular Drug Delivery Systems. Pharmaceutics 2021, 13, 60.
  14. Sellam, J.; Berenbaum, F. Is osteoarthritis a metabolic disease? Jt. Bone Spine 2013, 80, 568–573.
  15. Robinson, W.H.; Lepus, C.M.; Wang, Q.; Raghu, H.; Mao, R.; Lindstrom, T.M.; Sokolove, J. Low-grade inflammation as a key mediator of the pathogenesis of osteoarthritis. Nat. Rev. Rheumatol. 2016, 12, 580–592.
  16. Bannuru, R.R.; Osani, M.C.; Vaysbrot, E.E.; Arden, N.K.; Bennell, K.; Bierma-Zeinstra, S.M.A.; Kraus, V.B.; Lohmander, L.S.; Abbott, J.H.; Bhandari, M.; et al. OARSI guidelines for the non-surgical management of knee, hip, and polyarticular osteoarthritis. Osteoarthr. Cartil. 2019, 27, 1578–1589.
  17. Fuggle, N.R.; Cooper, C.; Oreffo, R.O.C.; Price, A.J.; Kaux, J.F.; Maheu, E.; Cutolo, M.; Honvo, G.; Conaghan, P.G.; Berenbaum, F.; et al. Alternative and complementary therapies in osteoarthritis and cartilage repair. Aging Clin. Exp. Res. 2020, 32, 547–560.
  18. Birke, H.; Kurita, G.P.; Sjogren, P.; Hojsted, J.; Simonsen, M.K.; Juel, K.; Ekholm, O. Chronic non-cancer pain and the epidemic prescription of opioids in the Danish population: Trends from 2000 to 2013. Acta Anaesthesiol. Scand. 2016, 60, 623–633.
  19. Pelletier, J.P.; Martel-Pelletier, J.; Rannou, F.; Cooper, C. Efficacy and safety of oral NSAIDs and analgesics in the management of osteoarthritis: Evidence from real-life setting trials and surveys. Semin. Arthritis Rheum. 2016, 45, S22–S27.
  20. Evans, C.H.; Kraus, V.B.; Setton, L.A. Progress in intra-articular therapy. Nat. Rev. Rheumatol. 2014, 10, 11–22.
  21. Maudens, P.; Jordan, O.; Allemann, E. Recent advances in intra-articular drug delivery systems for osteoarthritis therapy. Drug Discov. Today 2018, 23, 1761–1775.
  22. Rahnfeld, L.; Thamm, J.; Steiniger, F.; van Hoogevest, P.; Luciani, P. Study on the in situ aggregation of liposomes with negatively charged phospholipids for use as injectable depot formulation. Colloids Surfaces B 2018, 168, 10–17.
  23. Trif, M.; Roseanu, A.; Brock, J.H.; Brewer, J.M. Designing lipid nanostructures for local delivery of biologically active macromolecules. J. Liposome Res. 2007, 17, 237–248.
  24. Wehling, P.; Evans, C.; Wehling, J.; Maixner, W. Effectiveness of intra-articular therapies in osteoarthritis: A literature review. Ther. Adv. Musculoskelet. Dis. 2017, 9, 183–196.
  25. Karsdal, M.A.; Michaelis, M.; Ladel, C.; Siebuhr, A.S.; Bihlet, A.R.; Andersen, J.R.; Guehring, H.; Christiansen, C.; Bay-Jensen, A.C.; Kraus, V.B. Disease-modifying treatments for osteoarthritis (DMOADs) of the knee and hip: Lessons learned from failures and opportunities for the future. Osteoarthr. Cartil. 2016, 24, 2013–2021.
  26. Ahmed, S. Green tea polyphenol epigallocatechin 3-gallate in arthritis: Progress and promise. Arthritis Res. Ther. 2010, 12, 208.
  27. Akhtar, N.; Haqqi, T.M. Current nutraceuticals in the management of osteoarthritis: A review. Ther. Adv. Musculoskelet. Dis. 2012, 4, 181–207.
  28. Chin, K.Y. The spice for joint inflammation: Anti-inflammatory role of curcumin in treating osteoarthritis. Drug Des. Dev. Ther. 2016, 10, 3029–3042.
  29. Hochberg, M.C.; Martel-Pelletier, J.; Monfort, J.; Moller, I.; Castillo, J.R.; Arden, N.; Berenbaum, F.; Blanco, F.J.; Conaghan, P.G.; Domenech, G.; et al. Combined chondroitin sulfate and glucosamine for painful knee osteoarthritis: A multicentre, randomised, double-blind, non-inferiority trial versus celecoxib. Ann. Rheum. Dis. 2016, 75, 37–44.
  30. Kann, B.; Spengler, C.; Coradini, K.; Rigo, L.A.; Bennink, M.L.; Jacobs, K.; Offerhaus, H.L.; Beck, R.C.R.; Windbergs, M. Intracellular Delivery of Poorly Soluble Polyphenols: Elucidating the Interplay of Self-Assembling Nanocarriers and Human Chondrocytes. Anal. Chem. 2016, 88, 7014–7022.
  31. Lim, H.; Min, D.S.; Park, H.; Kim, H.P. Flavonoids interfere with NLRP3 inflammasome activation. Toxicol. Appl. Pharm. 2018, 355, 93–102.
  32. Nelson, K.M.; Dahlin, J.L.; Bisson, J.; Graham, J.; Pauli, G.F.; Walters, M.A. The Essential Medicinal Chemistry of Curcumin. J. Med. Chem. 2017, 60, 1620–1637.
  33. Reginster, J.Y.; Veronese, N. Highly purified chondroitin sulfate: A literature review on clinical efficacy and pharmacoeconomic aspects in osteoarthritis treatment. Aging Clin. Exp. Res. 2021, 33, 37–47.
  34. Trif, M.; Craciunescu, O. Liposomes as Delivery System of Chondroitin Sulfate to the Arthritic Joint by Intra-articular Administration. Austin Arthritis 2016, 1, 1011.
  35. Yagi, H.; Ulici, V.; Tuan, R.S. Polyphenols suppress inducible oxidative stress in human osteoarthritic and bovine chondrocytes. Osteoarthr. Cartil. Open 2020, 2, 100064.
  36. Burt, H.M.; Tsallas, A.; Gilchrist, S.; Liang, L.S. Intra-articular drug delivery systems: Overcoming the shortcomings of joint disease therapy. Expert Opin. Drug Deliv. 2009, 6, 17–26.
  37. Kavanaugh, T.E.; Werfel, T.A.; Cho, H.; Hasty, K.A.; Duvall, C.L. Particle-based technologies for osteoarthritis detection and therapy. Drug Deliv. Transl. Res. 2016, 6, 132–147.
  38. Ghafari, M.; Haghiralsadat, F.; Falahati-pour, S.K.; Reza, J.Z. Development of a novel liposomal nanoparticle formulation of cisplatin to breast cancer therapy. J. Cell Biochem. 2020, 121, 3584–3592.
  39. Allen, T.M.; Cullis, P.R. Liposomal drug delivery systems: From concept to clinical applications. Adv. Deliv. Rev. 2013, 65, 36–48.
  40. Beltrán-Gracia, E.; López-Camacho, A.; Higuera-Ciapara, I.; Velázquez-Fernández, J.B.; Vallejo-Cardona, A.A. Nanomedicine review: Clinical developments in liposomal applications. Cancer Nanotechnol. 2019, 10, 11.
  41. Cipollaro, L.; Trucillo, P.; Bragazzi, N.L.; Della Porta, G.; Reverchon, E.; Maffulli, N. Liposomes for Intra-Articular Analgesic Drug Delivery in Orthopedics: State-of-Art and Future Perspectives. Insights from a Systematic Mini-Review of the Literature. Medicina 2020, 56, 423.
  42. Pawar, V.A.; Manjappa, A.S.; Murumkar, P.R.; Gajaria, T.K.; Devkar, R.V.; Mishra, A.K.; Yadav, M.R. Drug-fortified liposomes as carriers for sustained release of NSAIDs: The concept and its validation in the animal model for the treatment of arthritis. Eur. J. Pharm. Sci. 2018, 125, 11–22.
  43. Gerwin, N.; Hops, C.; Lucke, A. Intraarticular drug delivery in osteoarthritis. Adv. Drug Deliv. Rev. 2006, 58, 226–242.
More